Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35217625

RESUMO

As natural chemokine inhibitors, evasin proteins produced in tick saliva are potential therapeutic agents for numerous inflammatory diseases. Engineering evasins to block the desired chemokines and avoid off-target side effects requires structural understanding of their target selectivity. Structures of the class A evasin EVA-P974 bound to human CC chemokine ligands 7 and 17 (CCL7 and CCL17) and to a CCL8-CCL7 chimera reveal that the specificity of class A evasins for chemokines of the CC subfamily is defined by conserved, rigid backbone-backbone interactions, whereas the preference for a subset of CC chemokines is controlled by side-chain interactions at four hotspots in flexible structural elements. Hotspot mutations alter target preference, enabling inhibition of selected chemokines. The structure of an engineered EVA-P974 bound to CCL2 reveals an underlying molecular mechanism of EVA-P974 target preference. These results provide a structure-based framework for engineering evasins as targeted antiinflammatory therapeutics.


Assuntos
Proteínas de Artrópodes/química , Quimiocinas/metabolismo , Inflamação/metabolismo , Engenharia de Proteínas , Carrapatos/metabolismo , Animais , Proteínas de Artrópodes/metabolismo , Ligação Proteica , Conformação Proteica , Receptores de Quimiocinas/metabolismo
2.
Nucleic Acids Res ; 49(5): 2403-2417, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33621982

RESUMO

TIA-1 is an RNA-binding protein that sequesters target RNA into stress granules under conditions of cellular stress. Promotion of stress granule formation by TIA-1 depends upon self-association of its prion-like domain that facilitates liquid-liquid phase separation and is thought to be enhanced via RNA binding. However, the mechanisms underlying the influence of RNA on TIA-1 self-association have not been previously demonstrated. Here we have investigated the self-associating properties of full-length TIA-1 in the presence of designed and native TIA-1 nucleic acid binding sites in vitro, monitoring phase separation, fibril formation and shape. We show that single stranded RNA and DNA induce liquid-liquid phase separation of TIA-1 in a multisite, sequence-specific manner and also efficiently promote formation of amyloid-like fibrils. Although RNA binding to a single site induces a small conformational change in TIA-1, this alone does not enhance phase separation of TIA-1. Tandem binding sites are required to enhance phase separation of TIA-1 and this is finely tuned by the protein:binding site stoichiometry rather than nucleic acid length. Native tandem TIA-1 binding sites within the 3' UTR of p53 mRNA also efficiently enhance phase separation of TIA-1 and thus may potentially act as potent nucleation sites for stress granule assembly.


Assuntos
RNA/metabolismo , Antígeno-1 Intracelular de Células T/química , Regiões 3' não Traduzidas , Amiloide/ultraestrutura , Sítios de Ligação , DNA/química , DNA/metabolismo , Humanos , Modelos Moleculares , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Conformação Proteica , RNA/química , Antígeno-1 Intracelular de Células T/metabolismo , Antígeno-1 Intracelular de Células T/ultraestrutura , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Nucleic Acids Res ; 48(14): 8006-8021, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32556302

RESUMO

The poliovirus type I IRES is able to recruit ribosomal machinery only in the presence of host factor PCBP2 that binds to stem-loop IV of the IRES. When PCBP2 is cleaved in its linker region by viral proteinase 3CD, translation initiation ceases allowing the next stage of replication to commence. Here, we investigate the interaction of PCBP2 with the apical region of stem-loop IV (SLIVm) of poliovirus RNA in its full-length and truncated form. CryoEM structure reconstruction of the full-length PCBP2 in complex with SLIVm solved to 6.1 Å resolution reveals a compact globular complex of PCBP2 interacting with the cruciform RNA via KH domains and featuring a prominent GNRA tetraloop. SEC-SAXS, SHAPE and hydroxyl-radical cleavage establish that PCBP2 stabilizes the SLIVm structure, but upon cleavage in the linker domain the complex becomes more flexible and base accessible. Limited proteolysis and REMSA demonstrate the accessibility of the linker region in the PCBP2/SLIVm complex and consequent loss of affinity of PCBP2 for the SLIVm upon cleavage. Together this study sheds light on the structural features of the PCBP2/SLIV complex vital for ribosomal docking, and the way in which this key functional interaction is regulated following translation of the poliovirus genome.


Assuntos
Iniciação Traducional da Cadeia Peptídica , Poliovirus/genética , RNA Viral/química , Proteínas de Ligação a RNA/química , Microscopia Crioeletrônica , Modelos Moleculares , Conformação de Ácido Nucleico , Conformação Proteica , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Espalhamento a Baixo Ângulo , Difração de Raios X
4.
Nucleic Acids Res ; 46(6): 3169-3186, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29346611

RESUMO

RIG-I (retinoic acid inducible gene-I) is a cytosolic innate immune protein that senses viral dsRNA with a 5'-triphosphate overhang. Upon interaction with dsRNA a de-repression of the RIG-I CARD domains takes place that ultimately leads to the production of type I interferons and pro-inflammatory cytokines. Here we investigate the RIG-I conformational rearrangement upon interaction with an activating 5'-triphosphate-10-base pair dsRNA hairpin loop (10bp) compared with a less active 5'-triphosphate-8-base pair dsRNA hairpin loop (8bp). We use size-exclusion chromatography-coupled small-angle X-ray scattering (SAXS) and limited tryptic digest experiments to show that that upon binding to 10 bp, but not 8 bp, RIG-I becomes extended and shows greater flexibility, reflecting the release of its CARDs. We also examined the effect of different ATP analogues on the conformational changes of RIG-I/dsRNA complexes. Of the analogues tested, the addition of ATP transition state analogue ADP-AlFx further assisted in the complete activation of RIG-I in complex with 10bp and also to some extent RIG-I bound to 8bp. Together these data provide solution-based evidence for the molecular mechanism of innate immune signaling by RIG-I as stimulated by short hairpin RNA and ATP.


Assuntos
Trifosfato de Adenosina/química , Proteína DEAD-box 58/química , Domínios Proteicos , RNA de Cadeia Dupla/química , Trifosfato de Adenosina/metabolismo , Sequência de Bases , Cromatografia em Gel , Proteína DEAD-box 58/metabolismo , Humanos , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Receptores Imunológicos , Espalhamento a Baixo Ângulo , Transdução de Sinais , Soluções/química , Difração de Raios X
5.
Mol Cell ; 41(4): 419-31, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21329880

RESUMO

Transcript-selective translational regulation of epithelial-mesenchymal transition (EMT) by transforming growth factor-ß (TGF-ß) is directed by the hnRNP E1-containing TGF-ß-activated-translational (BAT) mRNP complex. Herein, eukaryotic elongation factor-1 A1 (eEF1A1) is identified as an integral component of the BAT complex. Translational silencing of Dab2 and ILEI, two EMT transcripts, is mediated by the binding of hnRNP E1 and eEF1A1 to their 3'UTR BAT element, whereby hnRNP E1 stalls translational elongation by inhibiting the release of eEF1A1 from the ribosomal A site. TGF-ß-mediated hnRNP E1 phosphorylation, through Akt2, disrupts the BAT complex, thereby restoring translation of target EMT transcripts. Attenuation of hnRNP E1 expression in two noninvasive breast epithelial cells (NMuMG and MCF-7) not only induced EMT but also enabled cells to form metastatic lesions in vivo. Thus, translational regulation by TGF-ß at the elongation stage represents a critical checkpoint coordinating the expression of EMT transcripts required during development and in tumorigenesis and metastatic progression.


Assuntos
Neoplasias/genética , Elongação Traducional da Cadeia Peptídica/fisiologia , Ribonucleoproteínas/metabolismo , Animais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/fisiologia , Fator de Iniciação 1 em Eucariotos/genética , Fator de Iniciação 1 em Eucariotos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/metabolismo , Biossíntese de Peptídeos Independentes de Ácido Nucleico/fisiologia , Ribonucleoproteínas/genética , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
6.
Nucleic Acids Res ; 45(8): 4944-4957, 2017 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-28184449

RESUMO

TIA-1 (T-cell restricted intracellular antigen-1) is an RNA-binding protein involved in splicing and translational repression. It mainly interacts with RNA via its second and third RNA recognition motifs (RRMs), with specificity for U-rich sequences directed by RRM2. It has recently been shown that RRM3 also contributes to binding, with preferential binding for C-rich sequences. Here we designed UC-rich and CU-rich 10-nt sequences for engagement of both RRM2 and RRM3 and demonstrated that the TIA-1 RRM23 construct preferentially binds the UC-rich RNA ligand (5΄-UUUUUACUCC-3΄). Interestingly, this binding depends on the presence of Lys274 that is C-terminal to RRM3 and binding to equivalent DNA sequences occurs with similar affinity. Small-angle X-ray scattering was used to demonstrate that, upon complex formation with target RNA or DNA, TIA-1 RRM23 adopts a compact structure, showing that both RRMs engage with the target 10-nt sequences to form the complex. We also report the crystal structure of TIA-1 RRM2 in complex with DNA to 2.3 Šresolution providing the first atomic resolution structure of any TIA protein RRM in complex with oligonucleotide. Together our data support a specific mode of TIA-1 RRM23 interaction with target oligonucleotides consistent with the role of TIA-1 in binding RNA to regulate gene expression.


Assuntos
Proteínas de Ligação a DNA/química , DNA/química , Proteínas de Ligação a Poli(A)/química , Ribonucleosídeo Difosfato Redutase/química , Cristalografia por Raios X , DNA/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Humanos , Oligonucleotídeos/química , Proteínas de Ligação a Poli(A)/genética , Ligação Proteica/genética , Mapas de Interação de Proteínas/genética , Motivo de Reconhecimento de RNA/genética , Ribonucleosídeo Difosfato Redutase/genética , Antígeno-1 Intracelular de Células T
7.
Immunity ; 30(6): 777-88, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19464197

RESUMO

Ligation of the alphabeta T cell receptor (TCR) by a specific peptide-loaded major histocompatibility complex (pMHC) molecule initiates T cell signaling via the CD3 complex. However, the initial events that link antigen recognition to T cell signal transduction remain unclear. Here we show, via fluorescence-based experiments and structural analyses, that MHC-restricted antigen recognition by the alphabeta TCR results in a specific conformational change confined to the A-B loop within the alpha chain of the constant domain (Calpha). The apparent affinity constant of this A-B loop movement mirrored that of alphabeta TCR-pMHC ligation and was observed in two alphabeta TCRs with distinct pMHC specificities. The Ag-induced A-B loop conformational change could be inhibited by fixing the juxtapositioning of the constant domains and was shown to be reversible upon pMHC disassociation. Notably, the loop movement within the Calpha domain, although specific for an agonist pMHC ligand, was not observed with a pMHC antagonist. Moreover, mutagenesis of residues within the A-B loop impaired T cell signaling in an in vitro system of antigen-specific TCR stimulation. Collectively, our findings provide a basis for the earliest molecular events that underlie Ag-induced T cell triggering.


Assuntos
Antígenos/química , Receptores de Antígenos de Linfócitos T alfa-beta/química , Linfócitos T/imunologia , Animais , Antígenos/imunologia , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Mutação/genética , Peptídeos/química , Peptídeos/imunologia , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia
8.
Immunity ; 31(6): 897-908, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-20064448

RESUMO

T cells often alloreact with foreign human leukocyte antigens (HLA). Here we showed the LC13 T cell receptor (TCR), selected for recognition on self-HLA-B( *)0801 bound to a viral peptide, alloreacts with B44 allotypes (HLA-B( *)4402 and HLA-B( *)4405) bound to two different allopeptides. Despite extensive polymorphism between HLA-B( *)0801, HLA-B( *)4402, and HLA-B( *)4405 and the disparate sequences of the viral and allopeptides, the LC13 TCR engaged these peptide-HLA (pHLA) complexes identically, accommodating mimicry of the viral peptide by the allopeptide. The viral and allopeptides adopted similar conformations only after TCR ligation, revealing an induced-fit mechanism of molecular mimicry. The LC13 T cells did not alloreact against HLA-B( *)4403, and the single residue polymorphism between HLA-B( *)4402 and HLA-B( *)4403 affected the plasticity of the allopeptide, revealing that molecular mimicry was associated with TCR specificity. Accordingly, molecular mimicry that is HLA and peptide dependent is a mechanism for human T cell alloreactivity between disparate cognate and allogeneic pHLA complexes.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Antígenos HLA-B/imunologia , Mimetismo Molecular/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Linhagem Celular , Antígeno HLA-B8 , Humanos , Células Jurkat , Ativação Linfocitária/imunologia , Peptídeos/química , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Transfecção
9.
J Biol Chem ; 290(16): 10460-71, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25759384

RESUMO

The engagement of natural killer cell immunoglobulin-like receptors (KIRs) with their target ligands, human leukocyte antigen (HLA) molecules, is a critical component of innate immunity. Structurally, KIRs typically have either two (D1-D2) or three (D0-D1-D2) extracellular immunoglobulin domains, with the D1 and D2 domain recognizing the α1 and α2 helices of HLA, respectively, whereas the D0 domain of the KIR3DLs binds a loop region flanking the α1 helix of the HLA molecule. KIR2DL4 is distinct from other KIRs (except KIR2DL5) in that it does not contain a D1 domain and instead has a D0-D2 arrangement. Functionally, KIR2DL4 is also atypical in that, unlike all other KIRs, KIR2DL4 has both activating and inhibitory signaling domains. Here, we determined the 2.8 Å crystal structure of the extracellular domains of KIR2DL4. Structurally, KIR2DL4 is reminiscent of other KIR2DL receptors, with the D0 and D2 adopting the C2-type immunoglobulin fold arranged with an acute elbow angle. However, KIR2DL4 self-associated via the D0 domain in a concentration-dependent manner and was observed as a tetramer in the crystal lattice by size exclusion chromatography, dynamic light scattering, analytical ultracentrifugation, and small angle x-ray scattering experiments. The assignment of residues in the D0 domain to forming the KIR2DL4 tetramer precludes an interaction with HLA akin to that observed for KIR3DL1. Accordingly, no interaction was observed to HLA by direct binding studies. Our data suggest that the unique functional properties of KIR2DL4 may be mediated by self-association of the receptor.


Assuntos
Antígenos HLA-B/química , Antígenos HLA-G/química , Receptores KIR2DL4/química , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Antígenos HLA-B/genética , Antígenos HLA-B/metabolismo , Antígenos HLA-G/genética , Antígenos HLA-G/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mariposas/citologia , Mariposas/metabolismo , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores KIR2DL4/genética , Receptores KIR2DL4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
10.
Nature ; 467(7317): 844-8, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20944746

RESUMO

The pre-T-cell antigen receptor (pre-TCR), expressed by immature thymocytes, has a pivotal role in early T-cell development, including TCR ß-selection, survival and proliferation of CD4(-)CD8(-) double-negative thymocytes, and subsequent αß T-cell lineage differentiation. Whereas αßTCR ligation by the peptide-loaded major histocompatibility complex initiates T-cell signalling, pre-TCR-induced signalling occurs by means of a ligand-independent dimerization event. The pre-TCR comprises an invariant α-chain (pre-Tα) that pairs with any TCR ß-chain (TCRß) following successful TCR ß-gene rearrangement. Here we provide the basis of pre-Tα-TCRß assembly and pre-TCR dimerization. The pre-Tα chain comprised a single immunoglobulin-like domain that is structurally distinct from the constant (C) domain of the TCR α-chain; nevertheless, the mode of association between pre-Tα and TCRß mirrored that mediated by the Cα-Cß domains of the αßTCR. The pre-TCR had a propensity to dimerize in solution, and the molecular envelope of the pre-TCR dimer correlated well with the observed head-to-tail pre-TCR dimer. This mode of pre-TCR dimerization enabled the pre-Tα domain to interact with the variable (V) ß domain through residues that are highly conserved across the Vß and joining (J) ß gene families, thus mimicking the interactions at the core of the αßTCR's Vα-Vß interface. Disruption of this pre-Tα-Vß dimer interface abrogated pre-TCR dimerization in solution and impaired pre-TCR expression on the cell surface. Accordingly, we provide a mechanism of pre-TCR self-association that allows the pre-Tα chain to simultaneously 'sample' the correct folding of both the V and C domains of any TCR ß-chain, regardless of its ultimate specificity, which represents a critical checkpoint in T-cell development. This unusual dual-chaperone-like sensing function of pre-Tα represents a unique mechanism in nature whereby developmental quality control regulates the expression and signalling of an integral membrane receptor complex.


Assuntos
Multimerização Proteica , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/metabolismo , Cristalografia por Raios X , Rearranjo Gênico do Linfócito T/genética , Humanos , Modelos Moleculares , Mutação , Dobramento de Proteína , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T alfa-beta/química , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Transdução de Sinais , Soluções , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
11.
Proteins ; 83(7): 1225-37, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25287913

RESUMO

In this article, we describe the engineering and X-ray crystal structure of Thermal Green Protein (TGP), an extremely stable, highly soluble, non-aggregating green fluorescent protein. TGP is a soluble variant of the fluorescent protein eCGP123, which despite being highly stable, has proven to be aggregation-prone. The X-ray crystal structure of eCGP123, also determined within the context of this paper, was used to carry out rational surface engineering to improve its solubility, leading to TGP. The approach involved simultaneously eliminating crystal lattice contacts while increasing the overall negative charge of the protein. Despite intentional disruption of lattice contacts and introduction of high entropy glutamate side chains, TGP crystallized readily in a number of different conditions and the X-ray crystal structure of TGP was determined to 1.9 Å resolution. The structural reasons for the enhanced stability of TGP and eCGP123 are discussed. We demonstrate the utility of using TGP as a fusion partner in various assays and significantly, in amyloid assays in which the standard fluorescent protein, EGFP, is undesirable because of aberrant oligomerization.


Assuntos
Proteínas de Fluorescência Verde/química , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/química , Sequência de Aminoácidos , Amiloide/química , Bioensaio , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Temperatura Alta , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Estabilidade Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Eletricidade Estática
12.
Mol Microbiol ; 91(1): 110-20, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24261685

RESUMO

Protein biotinylation is catalysed by biotin protein ligase (BPL). The most characterized BPL is from Escherichia coli where it functions as both a biotin ligase and a homodimeric transcriptional repressor. Here we investigated another bifunctional BPL from the clinically important Staphylococcus aureus (SaBPL). Unliganded SaBPL (apo) exists in a dimer-monomer equilibrium at low micromolar concentrations - a stark contrast to E. coli BPL (EcBPL) that is monomeric under the same conditions. EMSA and SAXS analysis demonstrated that dimeric apo SaBPL adopted a conformation that was competent to bind DNA and necessary for it to function as a transcription factor. The SaBPL dimer-monomer dissociation constant was 5.8-fold tighter when binding the inhibitor biotin acetylene, but unchanged with biotin. F123, located in the dimer interface, was critical for homodimerization. Inhibition studies together with surface plasmon resonance analyses revealed a strong correlation between inhibitor potency and slow dissociation kinetics. A 24-fold difference in Ki values for these two enzymes was explained by differences in enzyme:inhibitor dissociation rates. Substitution of F123 in SaBPL and its equivalent in EcBPL altered both inhibitor potency and dissociation. Hence, F123 in SaBPL has novel roles in both protein dimerization and ligand-binding that have not been reported in EcBPL.


Assuntos
Sítios de Ligação/fisiologia , Biotina/metabolismo , Ligases/química , Ligases/metabolismo , Fenilalanina/metabolismo , Staphylococcus aureus/enzimologia , Motivos de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação/genética , Biotina/antagonistas & inibidores , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Ligantes , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Espalhamento a Baixo Ângulo , Staphylococcus aureus/genética , Ressonância de Plasmônio de Superfície , Difração de Raios X
13.
J Mol Recognit ; 28(5): 316-24, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25720550

RESUMO

The transfer of antibiotic resistance between bacteria is mediated by mobile genetic elements such as plasmids and transposons. TnpX is a member of the large serine recombinase subgroup of site-specific recombinases and is responsible for the excision and insertion of mobile genetic elements that encode chloramphenicol resistance in the pathogens Clostridium perfringens and Clostridium difficile. TnpX consists of three structural domains: domain I contains the catalytic site, whereas domains II and III contain DNA-binding motifs. We have solved the solution structure of residues 1-120 of the catalytic domain I of TnpX. The TnpX catalytic domain shares the same overall fold as other serine recombinases; however, differences are evident in the identity of the proposed hydrogen donor and in the size, amino acid composition, conformation, and dynamics of the TnpX active site loops. To obtain the interaction surface of TnpX1-120 , we titrated a DNA oligonucleotide containing the circular intermediate joint attCI recombination site into (15) N-labeled TnpX1-120 and observed progressive nuclear magnetic resonance chemical shift perturbations using (15) N HSQC spectra. Perturbations were largely confined to a region surrounding the catalytic serine and encompassed residues of the active site loops. Utilizing the perturbation map and the data-driven docking program, HADDOCK, we have generated a model of the DNA interaction complex for the TnpX catalytic domain.


Assuntos
Proteínas de Bactérias/química , DNA Bacteriano/química , Recombinases/química , Sequência de Aminoácidos , Domínio Catalítico , Clostridium perfringens/enzimologia , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Conformação de Ácido Nucleico , Ligação Proteica
14.
Nucleic Acids Res ; 41(5): 3436-45, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23325848

RESUMO

The retinoic acid inducible gene-I (RIG-I)-like family of receptors is positioned at the front line of our innate cellular defence system. RIG-I detects and binds to foreign duplex RNA in the cytoplasm of both immune and non-immune cells, and initiates the induction of type I interferons and pro-inflammatory cytokines. The mechanism of RIG-I activation by double-stranded RNA (dsRNA) involves a molecular rearrangement proposed to expose the N-terminal pair of caspase activation recruitment domains, enabling an interaction with interferon-beta promoter stimulator 1 (IPS-1) and thereby initiating downstream signalling. dsRNA is particularly stimulatory when longer than 20 bp, potentially through allowing binding of more than one RIG-I molecule. Here, we characterize full-length RIG-I and RIG-I subdomains combined with a stimulatory 29mer dsRNA using multi-angle light scattering and size-exclusion chromatography-coupled small-angle X-ray scattering, to build up a molecular model of RIG-I before and after the formation of a 2:1 protein:dsRNA assembly. We report the small-angle X-ray scattering-derived solution structure of the human apo-RIG-I and observe that on binding of RIG-I to dsRNA in a 2:1 ratio, the complex becomes highly extended and flexible. Hence, here we present the first model of the fully activated oligomeric RIG-I.


Assuntos
Apoproteínas/química , RNA Helicases DEAD-box/química , RNA de Cadeia Dupla/química , Cromatografia em Gel , Proteína DEAD-box 58 , Humanos , Modelos Moleculares , Fragmentos de Peptídeos/química , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteólise , Receptores Imunológicos , Espalhamento a Baixo Ângulo , Tripsina/química , Difração de Raios X
15.
Inorg Chem ; 53(20): 10996-1006, 2014 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-25271783

RESUMO

Both conventional solution-phase and direct solid-solid redox reactions between tetrathiafulvalene (TTF) and the vanadium-substituted polyoxometalate (n-Bu4N)3[SV(V)W11O40] give rise to microcrystalline or powdered semiconducting charge transfer solid material. A single-crystal X-ray structure derived from growing crystals from a MeCN-CH2Cl2 solution-phase redox reaction gives a stoichiometry of TTF4[SVW11O40]·2H2O·2CH2Cl2 and reveals that there are two crystallographically different TTF cation moieties based on (TTF2)(2+) dimers. While the color and morphology of the microcrystalline or powdered TTF4[SVW11O40] differ from the single crystals prepared for structural analysis, all materials are spectroscopically (infrared (IR), Raman with respect to the TTF bands, and electron paramagnetic resonance (EPR)) indistinguishable. Raman spectra suggest that the charge transfer is unevenly distributed across the (TTF2)(2+) dimers, which is postulated to give rise to enhanced mixed-valence features. Structural, spectral, and other properties, such as conductivity, are compared with results available on the recently published molybdenum TTF4[SVMo11O40]·2H2O·2CH2Cl2 analogue, where the charge distribution is uniform on all TTF cations. In both examples, the position of the V atom is located over several sites. Elemental analysis and voltammetric data also are consistent with the formulations deduced from structural and spectroscopic studies. The conductivity at room temperature is in the semiconducting range, but significantly greater than that for the Mo analogue. EPR spectra at temperatures down to the liquid helium regime confirm the presence of paramagnetic V(IV) and paramagnetic oxidized TTF. The newly isolated TTF-SV(IV)W11O40 material also has magnetic functionality derived from the cationic and anionic components.


Assuntos
Compostos Heterocíclicos/química , Molibdênio/química , Compostos Organometálicos/química , Compostos Organometálicos/síntese química , Compostos de Tungstênio/química , Vanádio/química , Cristalografia por Raios X , Técnicas Eletroquímicas , Modelos Moleculares , Oxirredução , Tamanho da Partícula , Propriedades de Superfície
16.
Nature ; 456(7222): 648-52, 2008 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-18971931

RESUMO

AB(5) toxins comprise an A subunit that corrupts essential eukaryotic cell functions, and pentameric B subunits that direct target-cell uptake after binding surface glycans. Subtilase cytotoxin (SubAB) is an AB(5) toxin secreted by Shiga toxigenic Escherichia coli (STEC), which causes serious gastrointestinal disease in humans. SubAB causes haemolytic uraemic syndrome-like pathology in mice through SubA-mediated cleavage of BiP/GRP78, an essential endoplasmic reticulum chaperone. Here we show that SubB has a strong preference for glycans terminating in the sialic acid N-glycolylneuraminic acid (Neu5Gc), a monosaccharide not synthesized in humans. Structures of SubB-Neu5Gc complexes revealed the basis for this specificity, and mutagenesis of key SubB residues abrogated in vitro glycan recognition, cell binding and cytotoxicity. SubAB specificity for Neu5Gc was confirmed using mouse tissues with a human-like deficiency of Neu5Gc and human cell lines fed with Neu5Gc. Despite lack of Neu5Gc biosynthesis in humans, assimilation of dietary Neu5Gc creates high-affinity receptors on human gut epithelia and kidney vasculature. This, and the lack of Neu5Gc-containing body fluid competitors in humans, confers susceptibility to the gastrointestinal and systemic toxicities of SubAB. Ironically, foods rich in Neu5Gc are the most common source of STEC contamination. Thus a bacterial toxin's receptor is generated by metabolic incorporation of an exogenous factor derived from food.


Assuntos
Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidade , Ácidos Neuramínicos/metabolismo , Polissacarídeos/química , Polissacarídeos/metabolismo , Subtilisinas/química , Subtilisinas/toxicidade , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular , Cristalografia por Raios X , Chaperona BiP do Retículo Endoplasmático , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Modelos Moleculares , Ácidos Neuramínicos/administração & dosagem , Ácidos Neuramínicos/farmacologia , Ligação Proteica , Subunidades Proteicas , Escherichia coli Shiga Toxigênica/química , Escherichia coli Shiga Toxigênica/patogenicidade , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Especificidade da Espécie , Especificidade por Substrato , Subtilisinas/genética , Subtilisinas/metabolismo , Análise de Sobrevida
17.
Nucleic Acids Res ; 40(11): 5101-14, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22344691

RESUMO

Poly-C-binding proteins are triple KH (hnRNP K homology) domain proteins with specificity for single stranded C-rich RNA and DNA. They play diverse roles in the regulation of protein expression at both transcriptional and translational levels. Here, we analyse the contributions of individual αCP1 KH domains to binding C-rich oligonucleotides using biophysical and structural methods. Using surface plasmon resonance (SPR), we demonstrate that KH1 makes the most stable interactions with both RNA and DNA, KH3 binds with intermediate affinity and KH2 only interacts detectibly with DNA. The crystal structure of KH1 bound to a 5'-CCCTCCCT-3' DNA sequence shows a 2:1 protein:DNA stoichiometry and demonstrates a molecular arrangement of KH domains bound to immediately adjacent oligonucleotide target sites. SPR experiments, with a series of poly-C-sequences reveals that cytosine is preferred at all four positions in the oligonucleotide binding cleft and that a C-tetrad binds KH1 with 10 times higher affinity than a C-triplet. The basis for this high affinity interaction is finally detailed with the structure determination of a KH1.W.C54S mutant bound to 5'-ACCCCA-3' DNA sequence. Together, these data establish the lead role of KH1 in oligonucleotide binding by αCP1 and reveal the molecular basis of its specificity for a C-rich tetrad.


Assuntos
Citosina/química , Ribonucleoproteínas Nucleares Heterogêneas/química , Oligonucleotídeos/química , Sítios de Ligação , DNA/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Modelos Moleculares , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/química , RNA Mensageiro/metabolismo
18.
Protein Sci ; 33(6): e4999, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38723106

RESUMO

Ticks produce chemokine-binding proteins, known as evasins, in their saliva to subvert the host's immune response. Evasins bind to chemokines and thereby inhibit the activation of their cognate chemokine receptors, thus suppressing leukocyte recruitment and inflammation. We recently described subclass A3 evasins, which, like other class A evasins, exclusively target CC chemokines but appear to use a different binding site architecture to control target selectivity among CC chemokines. We now describe the structural basis of chemokine recognition by the class A3 evasin EVA-ACA1001. EVA-ACA1001 binds to almost all human CC chemokines and inhibits receptor activation. Truncation mutants of EVA-ACA1001 showed that, unlike class A1 evasins, both the N- and C-termini of EVA-ACA1001 play minimal roles in chemokine binding. To understand the structural basis of its broad chemokine recognition, we determined the crystal structure of EVA-ACA1001 in complex with the human chemokine CCL16. EVA-ACA1001 forms backbone-backbone interactions with the CC motif of CCL16, a conserved feature of all class A evasin-chemokine complexes. A hydrophobic pocket in EVA-ACA1001, formed by several aromatic side chains and the unique disulfide bond of class A3 evasins, accommodates the residue immediately following the CC motif (the "CC + 1 residue") of CCL16. This interaction is shared with EVA-AAM1001, the only other class A3 evasins characterized to date, suggesting it may represent a common mechanism that accounts for the broad recognition of CC chemokines by class A3 evasins.


Assuntos
Modelos Moleculares , Humanos , Animais , Carrapatos/química , Carrapatos/metabolismo , Cristalografia por Raios X , Sítios de Ligação , Proteínas de Artrópodes/química , Proteínas de Artrópodes/metabolismo , Proteínas de Artrópodes/genética , Ligação Proteica , Quimiocinas/química , Quimiocinas/metabolismo , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/metabolismo
19.
Biochemistry ; 52(48): 8652-62, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24175947

RESUMO

The cocaine-binding aptamer is unusual in that it tightly binds molecules other than the ligand it was selected for. Here, we study the interaction of the cocaine-binding aptamer with one of these off-target ligands, quinine. Isothermal titration calorimetry was used to quantify the quinine-binding affinity and thermodynamics of a set of sequence variants of the cocaine-binding aptamer. We find that the affinity of the cocaine-binding aptamer for quinine is 30-40 times stronger than it is for cocaine. Competitive-binding studies demonstrate that both quinine and cocaine bind at the same site on the aptamer. The ligand-induced structural-switching binding mechanism of an aptamer variant that contains three base pairs in stem 1 is retained with quinine as a ligand. The short stem 1 aptamer is unfolded or loosely folded in the free form and becomes folded when bound to quinine. This folding is confirmed by NMR spectroscopy and by the short stem 1 construct having a more negative change in heat capacity of quinine binding than is seen when stem 1 has six base pairs. Small-angle X-ray scattering (SAXS) studies of the free aptamer and both the quinine- and the cocaine-bound forms show that, for the long stem 1 aptamers, the three forms display similar hydrodynamic properties, and the ab initio shape reconstruction structures are very similar. For the short stem 1 aptamer there is a greater variation among the SAXS-derived ab initio shape reconstruction structures, consistent with the changes expected with its structural-switching binding mechanism.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Cocaína/metabolismo , Quinina/metabolismo , Aptâmeros de Nucleotídeos/química , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Cocaína/química , Hidrodinâmica , Ligantes , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Concentração Osmolar , Quinina/química , Especificidade por Substrato , Termodinâmica
20.
J Biol Chem ; 287(21): 17823-17832, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22437830

RESUMO

There is a well documented need to replenish the antibiotic pipeline with new agents to combat the rise of drug resistant bacteria. One strategy to combat resistance is to discover new chemical classes immune to current resistance mechanisms that inhibit essential metabolic enzymes. Many of the obvious drug targets that have no homologous isozyme in the human host have now been investigated. Bacterial drug targets that have a closely related human homologue represent a new frontier in antibiotic discovery. However, to avoid potential toxicity to the host, these inhibitors must have very high selectivity for the bacterial enzyme over the human homolog. We have demonstrated that the essential enzyme biotin protein ligase (BPL) from the clinically important pathogen Staphylococcus aureus could be selectively inhibited. Linking biotin to adenosine via a 1,2,3 triazole yielded the first BPL inhibitor selective for S. aureus BPL over the human equivalent. The synthesis of new biotin 1,2,3-triazole analogues using click chemistry yielded our most potent structure (K(i) 90 nM) with a >1100-fold selectivity for the S. aureus BPL over the human homologue. X-ray crystallography confirmed the mechanism of inhibitor binding. Importantly, the inhibitor showed cytotoxicity against S. aureus but not cultured mammalian cells. The biotin 1,2,3-triazole provides a novel pharmacophore for future medicinal chemistry programs to develop this new antibiotic class.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Biotina , Farmacorresistência Bacteriana/efeitos dos fármacos , Inibidores Enzimáticos , Ligases/antagonistas & inibidores , Staphylococcus aureus/enzimologia , Triazóis , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Biotina/química , Biotina/farmacologia , Linhagem Celular , Química Click , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Ligases/química , Ligases/metabolismo , Ligação Proteica , Triazóis/síntese química , Triazóis/química , Triazóis/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA